Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nat Commun ; 10(1): 4910, 2019 10 28.
Article in English | MEDLINE | ID: mdl-31659163

ABSTRACT

AspH is an endoplasmic reticulum (ER) membrane-anchored 2-oxoglutarate oxygenase whose C-terminal oxygenase and tetratricopeptide repeat (TPR) domains present in the ER lumen. AspH catalyses hydroxylation of asparaginyl- and aspartyl-residues in epidermal growth factor-like domains (EGFDs). Here we report crystal structures of human AspH, with and without substrate, that reveal substantial conformational changes of the oxygenase and TPR domains during substrate binding. Fe(II)-binding by AspH is unusual, employing only two Fe(II)-binding ligands (His679/His725). Most EGFD structures adopt an established fold with a conserved Cys1-3, 2-4, 5-6 disulfide bonding pattern; an unexpected Cys3-4 disulfide bonding pattern is observed in AspH-EGFD substrate complexes, the catalytic relevance of which is supported by studies involving stable cyclic peptide substrate analogues and by effects of Ca(II) ions on activity. The results have implications for EGFD disulfide pattern processing in the ER and will enable medicinal chemistry efforts targeting human 2OG oxygenases.


Subject(s)
Calcium-Binding Proteins/chemistry , Membrane Proteins/chemistry , Mixed Function Oxygenases/chemistry , Muscle Proteins/chemistry , Amino Acid Sequence , Asparagine/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Catalytic Domain , Crystallography , Disulfides/chemistry , Disulfides/metabolism , Epidermal Growth Factor/metabolism , Ferrous Compounds/chemistry , Ferrous Compounds/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Protein Conformation
2.
Nat Methods ; 15(3): 201-206, 2018 03.
Article in English | MEDLINE | ID: mdl-29334379

ABSTRACT

Sequencing the RNA in a biological sample can unlock a wealth of information, including the identity of bacteria and viruses, the nuances of alternative splicing or the transcriptional state of organisms. However, current methods have limitations due to short read lengths and reverse transcription or amplification biases. Here we demonstrate nanopore direct RNA-seq, a highly parallel, real-time, single-molecule method that circumvents reverse transcription or amplification steps. This method yields full-length, strand-specific RNA sequences and enables the direct detection of nucleotide analogs in RNA.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Nanopores , RNA, Fungal/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Sequence Analysis, RNA/methods
3.
Chemistry ; 23(52): 12815-12824, 2017 Sep 18.
Article in English | MEDLINE | ID: mdl-28703303

ABSTRACT

Isopenicillin N synthase (IPNS) catalyses the four-electron oxidation of a tripeptide, l-δ-(α-aminoadipoyl)-l-cysteinyl-d-valine (ACV), to give isopenicillin N (IPN), the first-formed ß-lactam in penicillin and cephalosporin biosynthesis. IPNS catalysis is dependent upon an iron(II) cofactor and oxygen as a co-substrate. In the absence of substrate, the carbonyl oxygen of the side-chain amide of the penultimate residue, Gln330, co-ordinates to the active-site metal iron. Substrate binding ablates the interaction between Gln330 and the metal, triggering rearrangement of seven C-terminal residues, which move to take up a conformation that extends the final α-helix and encloses ACV in the active site. Mutagenesis studies are reported, which probe the role of the C-terminal and other aspects of the substrate binding pocket in IPNS. The hydrophobic nature of amino acid side-chains around the ACV binding pocket is important in catalysis. Deletion of seven C-terminal residues exposes the active site and leads to formation of a new type of thiol oxidation product. The isolated product is shown by LC-MS and NMR analyses to be the ene-thiol tautomer of a dithioester, made up from two molecules of ACV linked between the thiol sulfur of one tripeptide and the oxidised cysteinyl ß-carbon of the other. A mechanism for its formation is proposed, supported by an X-ray crystal structure, which shows the substrate ACV bound at the active site, its cysteinyl ß-carbon exposed to attack by a second molecule of substrate, adjacent. Formation of this product constitutes a new mode of reaction for IPNS and non-heme iron oxidases in general.


Subject(s)
Aldehydes/metabolism , Esters/metabolism , Oxidoreductases/metabolism , Sulfhydryl Compounds/chemistry , Aldehydes/chemistry , Binding Sites , Biocatalysis , Catalytic Domain , Cephalosporins/biosynthesis , Cephalosporins/chemistry , Chromatography, High Pressure Liquid , Crystallography, X-Ray , Esters/chemistry , Iron/chemistry , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Conformation , Mutagenesis , Oxidation-Reduction , Oxidoreductases/genetics , Oxygen/chemistry , Oxygen/metabolism , Penicillins/biosynthesis , Penicillins/chemistry , Substrate Specificity
4.
Nat Commun ; 7: 11598, 2016 05 12.
Article in English | MEDLINE | ID: mdl-27176125

ABSTRACT

The invertebrate cytolysin lysenin is a member of the aerolysin family of pore-forming toxins that includes many representatives from pathogenic bacteria. Here we report the crystal structure of the lysenin pore and provide insights into its assembly mechanism. The lysenin pore is assembled from nine monomers via dramatic reorganization of almost half of the monomeric subunit structure leading to a ß-barrel pore ∼10 nm long and 1.6-2.5 nm wide. The lysenin pore is devoid of additional luminal compartments as commonly found in other toxin pores. Mutagenic analysis and atomic force microscopy imaging, together with these structural insights, suggest a mechanism for pore assembly for lysenin. These insights are relevant to the understanding of pore formation by other aerolysin-like pore-forming toxins, which often represent crucial virulence factors in bacteria.


Subject(s)
Cytotoxins/chemistry , Cytotoxins/metabolism , Invertebrates/chemistry , Animals , Crystallography, X-Ray , Microscopy, Atomic Force , Porosity , Protein Structure, Secondary , Toxins, Biological/chemistry
5.
Org Biomol Chem ; 11(5): 732-745, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23151668

ABSTRACT

Inhibition of the hypoxia-inducible factor (HIF) prolyl hydroxylases (PHD or EGLN enzymes) is of interest for the treatment of anemia and ischemia-related diseases. Most PHD inhibitors work by binding to the single ferrous ion and competing with 2-oxoglutarate (2OG) co-substrate for binding at the PHD active site. Non-specific iron chelators also inhibit the PHDs, both in vitro and in cells. We report the identification of dual action PHD inhibitors, which bind to the active site iron and also induce the binding of a second iron ion at the active site. Following analysis of small-molecule iron complexes and application of non-denaturing protein mass spectrometry to assess PHD2·iron·inhibitor stoichiometry, selected diacylhydrazines were identified as PHD2 inhibitors that induce the binding of a second iron ion. Some compounds were shown to inhibit the HIF hydroxylases in human hepatoma and renal carcinoma cell lines.


Subject(s)
Hydrazines/chemistry , Hydrazines/pharmacology , Iron/metabolism , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Procollagen-Proline Dioxygenase/metabolism , Catalytic Domain , Cell Line, Tumor , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases , Molecular Docking Simulation , Procollagen-Proline Dioxygenase/chemistry , Protein Binding/drug effects , Spectrometry, Mass, Electrospray Ionization
6.
Bioorg Med Chem Lett ; 20(20): 6125-8, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20822901

ABSTRACT

Aromatic analogues of the 2-oxoglutarate co-substrate of the hypoxia-inducible factor hydroxylases are shown to bind at the active site iron: Pyridine-2,4-dicarboxylate binds as anticipated with a single molecule chelating the iron in a bidentate manner. The binding mode of a hydroxamic acid analogue, at least in the crystalline state, is unusual because two molecules of the inhibitor are observed at the active site and partial displacement of the iron binding aspartyl residue was observed.


Subject(s)
Ketoglutaric Acids/chemistry , Ketoglutaric Acids/pharmacology , Repressor Proteins/metabolism , Binding Sites , Catalytic Domain , Humans , Mixed Function Oxygenases , Models, Molecular , Protein Binding , Repressor Proteins/chemistry
7.
Angew Chem Int Ed Engl ; 48(10): 1784-7, 2009.
Article in English | MEDLINE | ID: mdl-19180614

ABSTRACT

How PHDs achieve specificity: trans-4-prolyl hydroxylation of the transcription factor HIF occurs with stereochemical retention. Substrate-analogue studies show how the von Hippel Lindau tumor suppressor protein (pVHL) and the oxygen-sensing hydroxylases (PHDs) achieve specificity for hydroxyprolyl/prolyl residues for the C(4)-exo/endo prolyl conformations, respectively.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Oxygen/metabolism , Procollagen-Proline Dioxygenase/metabolism , Biosensing Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases , Protein Conformation , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stereoisomerism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
8.
Science ; 318(5855): 1469-72, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17991826

ABSTRACT

Variants in the FTO (fat mass and obesity associated) gene are associated with increased body mass index in humans. Here, we show by bioinformatics analysis that FTO shares sequence motifs with Fe(II)- and 2-oxoglutarate-dependent oxygenases. We find that recombinant murine Fto catalyzes the Fe(II)- and 2OG-dependent demethylation of 3-methylthymine in single-stranded DNA, with concomitant production of succinate, formaldehyde, and carbon dioxide. Consistent with a potential role in nucleic acid demethylation, Fto localizes to the nucleus in transfected cells. Studies of wild-type mice indicate that Fto messenger RNA (mRNA) is most abundant in the brain, particularly in hypothalamic nuclei governing energy balance, and that Fto mRNA levels in the arcuate nucleus are regulated by feeding and fasting. Studies can now be directed toward determining the physiologically relevant FTO substrate and how nucleic acid methylation status is linked to increased fat mass.


Subject(s)
DNA/metabolism , Ketoglutaric Acids/metabolism , Oxo-Acid-Lyases/genetics , Oxo-Acid-Lyases/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Amino Acid Sequence , Animals , Brain/enzymology , Brain/metabolism , Cell Nucleus/enzymology , Computational Biology , DNA Methylation , DNA, Single-Stranded/metabolism , Eating , Energy Metabolism , Fasting , Ferrous Compounds/metabolism , Hypothalamus/enzymology , Hypothalamus/metabolism , Male , Mice , Mixed Function Oxygenases , Molecular Sequence Data , Oxo-Acid-Lyases/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Succinic Acid/metabolism , Thymine/analogs & derivatives , Thymine/metabolism
9.
J Biol Chem ; 282(5): 3293-301, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17135241

ABSTRACT

In humans both the levels and activity of the alpha-subunit of the hypoxia-inducible transcription factor (HIF-alpha) are regulated by its post-translation hydroxylation as catalyzed by iron- and 2-oxoglutarate (2OG)-dependent prolyl and asparaginyl hydroxylases (PHD1-3 and factor-inhibiting HIF (FIH), respectively). One consequence of hypoxia is the accumulation of tricarboxylic acid cycle intermediates (TCAIs). In vitro assays were used to assess non-2OG TCAIs as inhibitors of purified PHD2 and FIH. Under the assay conditions, no significant FIH inhibition was observed by the TCAIs or pyruvate, but fumarate, succinate, and isocitrate inhibited PHD2. Mass spectrometric analyses under nondenaturing conditions were used to investigate the binding of TCAIs to PHD2 and supported the solution studies. X-ray crystal structures of FIH in complex with Fe(II) and fumarate or succinate revealed similar binding modes for each in the 2OG co-substrate binding site. The in vitro results suggest that the cellular inhibition of PHD2, but probably not FIH, by fumarate and succinate may play a role in the Warburg effect providing that appropriate relative concentrations of the components are achieved under physiological conditions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Citric Acid Cycle , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mixed Function Oxygenases/metabolism , Protein-Lysine 6-Oxidase/metabolism , Breast/enzymology , Breast/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Placental Lactogen/metabolism , Protein-Lysine 6-Oxidase/genetics
10.
Proc Natl Acad Sci U S A ; 103(26): 9814-9, 2006 Jun 27.
Article in English | MEDLINE | ID: mdl-16782814

ABSTRACT

Cellular and physiological responses to changes in dioxygen levels in metazoans are mediated via the posttranslational oxidation of hypoxia-inducible transcription factor (HIF). Hydroxylation of conserved prolyl residues in the HIF-alpha subunit, catalyzed by HIF prolyl-hydroxylases (PHDs), signals for its proteasomal degradation. The requirement of the PHDs for dioxygen links changes in dioxygen levels with the transcriptional regulation of the gene array that enables the cellular response to chronic hypoxia; the PHDs thus act as an oxygen-sensing component of the HIF system, and their inhibition mimics the hypoxic response. We describe crystal structures of the catalytic domain of human PHD2, an important prolyl-4-hydroxylase in the human hypoxic response in normal cells, in complex with Fe(II) and an inhibitor to 1.7 A resolution. PHD2 crystallizes as a homotrimer and contains a double-stranded beta-helix core fold common to the Fe(II) and 2-oxoglutarate-dependant dioxygenase family, the residues of which are well conserved in the three human PHD enzymes (PHD 1-3). The structure provides insights into the hypoxic response, helps to rationalize a clinically observed mutation leading to familial erythrocytosis, and will aid in the design of PHD selective inhibitors for the treatment of anemia and ischemic disease.


Subject(s)
Catalytic Domain , Oxygen/metabolism , Procollagen-Proline Dioxygenase/chemistry , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Procollagen-Proline Dioxygenase/genetics , Protein Conformation , von Hippel-Lindau Disease/genetics
12.
FEBS Lett ; 579(23): 5170-4, 2005 Sep 26.
Article in English | MEDLINE | ID: mdl-16153644

ABSTRACT

The ferrous iron and 2-oxoglutarate (2OG) dependent oxygenases catalyse two electron oxidation reactions by coupling the oxidation of substrate to the oxidative decarboxylation of 2OG, giving succinate and carbon dioxide coproducts. The evidence available on the level of incorporation of one atom from dioxygen into succinate is inconclusive. Here, we demonstrate that five members of the 2OG oxygenase family, AlkB from Escherichia coli, anthocyanidin synthase and flavonol synthase from Arabidopsis thaliana, and prolyl hydroxylase domain enzyme 2 and factor inhibiting hypoxia-inducible factor-1 from Homo sapiens all incorporate a single oxygen atom, almost exclusively derived from dioxygen, into the succinate co-product.


Subject(s)
Bacterial Proteins/metabolism , Iron/metabolism , Ketoglutaric Acids/metabolism , Oxygen/metabolism , Oxygenases/metabolism , Plant Proteins/metabolism , Succinic Acid/metabolism , Bacterial Proteins/chemistry , Humans , Iron/chemistry , Ketoglutaric Acids/chemistry , Molecular Structure , Oxidation-Reduction , Oxygen/chemistry , Oxygen Isotopes/chemistry , Oxygen Isotopes/metabolism , Oxygenases/chemistry , Plant Proteins/chemistry , Succinic Acid/chemistry
13.
J Am Chem Soc ; 127(21): 7680-1, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15913349

ABSTRACT

A set of four non-heme iron(II) and 2-oxoglutarate-dependent enzymes catalyze the post-translational modification of a transcription factor, hypoxia inducible factor (HIF), that mediates the hypoxic response in animals. Hydroxylation of HIF both causes its degradation and limits its activity. We describe how the use of structural data coupled to solid-phase synthesis led to the discovery of a selective inhibitor of one of the HIF hydroxylases. The inhibitor N-oxalyl-d-phenylalanine was shown to inhibit the HIF asparaginyl hydroxylase (FIH) but not a HIF prolyl hydroxylase. A crystal structure of the inhibitor complexed to FIH reveals that it binds in the 2OG and, likely, in the dioxygen binding site. The results will help to enable the modulation of the hypoxic response for the up-regulation of specific genes of biomedical importance, such as erythropoietin and vascular endothelial growth factor.


Subject(s)
Procollagen-Proline Dioxygenase/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Kinetics , Mixed Function Oxygenases , Models, Molecular , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Phenylalanine/analogs & derivatives , Phenylalanine/metabolism , Procollagen-Proline Dioxygenase/chemistry , Procollagen-Proline Dioxygenase/metabolism , Protein Structure, Secondary , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
14.
J Am Chem Soc ; 127(2): 506-7, 2005 Jan 19.
Article in English | MEDLINE | ID: mdl-15643858

ABSTRACT

Chlorination-elimination chemistry coupled with three-component Joullié-Ugi reaction and facile deprotection allowed efficient access to an array of polyhydroxylated pyrrolidines through parallel synthesis that may be considered to be a library of imino (aza) sugars (glycomimetics) and/or dihydroxyprolyl peptides (peptidomimetics). The utility of generating such a library was illustrated by screening against 15 different targets that revealed potent and selective inhibition of the Gaucher's disease glycosyltransferase enzyme glucosylceramide synthase and of primary pathogen model for human hepatitis C virus (HCV) and bovine diarrhoeal virus (BVDV). An observed selectivity for this HCV model over hepatitis B virus and remarkably low toxicity suggest a novel mode of action.


Subject(s)
Antiviral Agents/chemistry , Biomimetic Materials/chemistry , Glycopeptides/chemistry , Pyrrolidines/chemistry , Antiviral Agents/pharmacology , Aza Compounds/chemistry , Aza Compounds/pharmacology , Biomimetic Materials/pharmacology , Carbohydrates/chemistry , Carbohydrates/pharmacology , Diarrhea Viruses, Bovine Viral/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Erythritol/chemistry , Erythritol/pharmacology , Glycopeptides/pharmacology , Glycoside Hydrolases/antagonists & inhibitors , Hepatitis B virus/drug effects , Hydroxyproline/analogs & derivatives , Hydroxyproline/pharmacology , Pyrrolidines/pharmacology , Sugar Alcohols/chemistry , Sugar Alcohols/pharmacology
15.
Mol Biosyst ; 1(4): 321-4, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16880998

ABSTRACT

Regulation of the hypoxic response in humans is regulated by the post-translational hydroxylation of hypoxia inducible transcription factor; a recombinant form of a human prolyl-4-hydroxylase (PHD2) was characterised and shown to have an unexpectedly high affinity for, and to copurify with endogenous levels of, its Fe(ii) cofactor and 2-oxoglutarate cosubstrate.


Subject(s)
Ferrous Compounds/chemistry , Hypoxia-Inducible Factor 1/chemistry , Ketoglutaric Acids/chemistry , Procollagen-Proline Dioxygenase/chemistry , Binding Sites , Chromatography, Liquid , Humans , Mass Spectrometry , Models, Molecular
16.
Biochem J ; 383(Pt. 3): 429-37, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15239670

ABSTRACT

HIF (hypoxia-inducible factor) is an alphabeta transcription factor that modulates the hypoxic response in many animals. The cellular abundance and activity of HIF-alpha are regulated by its post-translational hydroxylation. The hydroxylation of HIF is catalysed by PHD (prolyl hydroxylase domain) enzymes and FIH (factorinhibiting HIF), all of which are 2-oxoglutarate- and Fe(II)-dependent dioxygenases. FIH hydroxylates a conserved asparagine residue in HIF-alpha (Asn-803), which blocks the binding of HIF to the transcriptional co-activator p300, preventing transcription of hypoxia-regulated genes under normoxic conditions. In the present paper, we report studies on possible mechanisms for the regulation of FIH activity. Recently solved crystal structures of FIH indicate that it is homodimeric. Site-directed mutants of FIH at residues Leu-340 and Ile-344, designed to disrupt dimerization, were generated in order to examine the importance of the dimeric state in determining FIH activity. A single point mutant, L340R (Leu-340-->Arg), was shown to be predominantly monomeric and to have lost catalytic activity as measured by assays monitoring 2-oxoglutarate turnover and asparagine hydroxylation. In contrast, the I344R (Ile-344-->Arg) mutant was predominantly dimeric and catalytically active. The results imply that the homodimeric form of FIH is required for productive substrate binding. The structural data also revealed a hydrophobic interaction formed between FIH and a conserved leucine residue (Leu-795) on the HIF substrate, which is close to the dimer interface. A recent report has revealed that phosphorylation of Thr-796, which is adjacent to Leu-795, enhances the transcriptional response in hypoxia. Consistent with this, we show that phosphorylation of Thr-796 prevents the hydroxylation of Asn-803 by FIH.


Subject(s)
Transcription Factors/antagonists & inhibitors , Amino Acid Sequence , Amino Acid Substitution/genetics , Amino Acid Substitution/physiology , Animals , Arginine/genetics , Arginine/physiology , Catalytic Domain/genetics , Catalytic Domain/physiology , Dimerization , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Leucine/genetics , Leucine/physiology , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed/genetics , Mutagenesis, Site-Directed/physiology , Mutation, Missense/genetics , Mutation, Missense/physiology , Peptides/chemical synthesis , Peptides/chemistry , Peptides/metabolism , Peptides/physiology , Phosphorylation , Point Mutation/genetics , Rats , Spectrometry, Mass, Electrospray Ionization/methods , Substrate Specificity/genetics , Substrate Specificity/physiology , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/physiology , Xenopus Proteins/chemistry , Zebrafish Proteins/chemistry
17.
Bioorg Med Chem Lett ; 13(16): 2677-80, 2003 Aug 18.
Article in English | MEDLINE | ID: mdl-12873492

ABSTRACT

Hydroxylation of hypoxia-inducible factor, a nuclear transcription factor, is catalysed by iron and 2-oxoglutarate dependent hydroxylases. Various analogues of the 2-oxoglutarate cosubstrate were synthesised and shown to inhibit the activity of human hypoxia-inducible factor-1alpha prolyl hydroxylases in cell-free extracts.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Ketoglutaric Acids/chemical synthesis , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Transcription Factors/metabolism , Catalysis , Cell-Free System , Enzyme Inhibitors/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Iron/chemistry , Iron Chelating Agents/pharmacology , Isoenzymes/antagonists & inhibitors , Ketoglutaric Acids/pharmacology , Molecular Structure
19.
J Biol Chem ; 278(12): 10157-61, 2003 Mar 21.
Article in English | MEDLINE | ID: mdl-12517755

ABSTRACT

AlkB is one of four proteins involved in the adaptive response to DNA alkylation damage in Escherichia coli and is highly conserved from bacteria to humans. Recent analyses have verified the prediction that AlkB is a member of the Fe(II) and 2-oxoglutarate (2OG)-dependent oxygenase family of enzymes. AlkB mediates repair of methylated DNA by direct demethylation of 1-methyladenine and 3-methylcytosine lesions. Other members of the Fe(II) and 2OG-dependent oxygenase family, including those involved in the hypoxic response, are targets for therapeutic intervention. Assays measuring 2OG turnover were used to investigate the selectivity of AlkB. 1-Methyladenosine, 1-methyl-2'-deoxyadenosine, 3-methylcytidine, and 3-methyl-2'-deoxycytidine all stimulated 2OG turnover by AlkB but were not demethylated indicating an uncoupling of 2OG and prime substrate oxidation and that oligomeric DNA is required for hydroxylation and subsequent demethylation. In contrast the equivalent unmethylated nucleosides did not stimulate 2OG turnover indicating that the presence of a methyl group in the substrate is important in initiating oxidation of 2OG. Stimulation of 2OG turnover by 1-methyladenosine was highly dependent on the presence of a reducing agent, ascorbate or dithiothreitol. Following the observation that AlkB is inhibited by high concentrations of 2OG, analogues of 2OG, including 2-mercaptoglutarate, were found to specifically inhibit AlkB. The flavonoid quercetin inhibits both AlkB and the 2OG oxygenase factor-inhibiting hypoxia-inducible factor (FIH) in vitro. FIH inhibition by quercetin occurs in the presence of excess iron indicating a specific interaction, while the inhibition of AlkB by quercetin is, predominantly, due to nonspecific iron chelation.


Subject(s)
Escherichia coli Proteins/antagonists & inhibitors , Mixed Function Oxygenases/antagonists & inhibitors , Ascorbic Acid/pharmacology , Escherichia coli Proteins/metabolism , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit , Ketoglutaric Acids/metabolism , Mixed Function Oxygenases/metabolism , Quercetin/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
20.
J Biol Chem ; 278(3): 1802-6, 2003 Jan 17.
Article in English | MEDLINE | ID: mdl-12446723

ABSTRACT

The activity of the transcription factor hypoxia-inducible factor (HIF) is regulated by oxygen-dependent hydroxylation. Under normoxic conditions, hydroxylation of proline residues triggers destruction of its alpha-subunit while hydroxylation of Asn(803) in the C-terminal transactivation domain of HIF-1 alpha (CAD) prevents its interaction with p300. Here we report crystal structures of the asparagine hydroxylase (factor-inhibiting HIF, FIH) complexed with Fe((II)), 2-oxoglutarate cosubstrate, and CAD fragments, which reveal the structural basis of HIF modification. CAD binding to FIH occurs via an induced fit process at two distinct interaction sites. At the hydroxylation site CAD adopts a loop conformation, contrasting with a helical conformation for the same residues when bound to p300. Asn(803) of CAD is buried and precisely orientated in the active site such that hydroxylation occurs at its beta-carbon. Together with structures with the inhibitors Zn((II)) and N-oxaloylglycine, analysis of the FIH-CAD complexes will assist design of hydroxylase inhibitors with proangiogenic properties. Conserved structural motifs within FIH imply it is one of an extended family of Fe((II)) oxygenases involved in gene regulation.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors , Amino Acid Sequence , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , Hypoxia-Inducible Factor 1 , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/chemistry , Oxidation-Reduction , Protein Conformation , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...